1
|
Azbazdar Y, De Robertis EM. Double assurance in the induction of axial development by egg dorsal determinants in Xenopus embryos. Proc Natl Acad Sci U S A 2025; 122:e2421772122. [PMID: 39928870 PMCID: PMC11848351 DOI: 10.1073/pnas.2421772122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/04/2025] [Indexed: 02/12/2025] Open
Abstract
We recently reported that microinjection of Xenopus nodal-related (xnr) mRNAs into β-catenin-depleted Xenopus embryos rescued a complete dorsal axis. Xnrs mediate the signal of the Nieuwkoop center that induces the Spemann-Mangold organizer in the overlying mesoderm, a process inhibited by the Nodal antagonist Cerberus-short (CerS). However, β-catenin also induces a second signaling center in the dorsal prospective ectoderm, designated the Blastula Chordin and Noggin Expression (BCNE) center, in which the homeobox gene siamois (sia) plays a major role. In this study, we asked whether the Xnrs and Sia depend on each other or function on parallel pathways. Expression of both genes induced β-catenin-depleted embryos to form complete axes with heads and eyes via the activation of similar sets of downstream organizer-specific genes. Xnrs did not activate siamois, and, conversely, Sia did not activate xnrs, although both were induced by β-catenin stabilization. Depletion with morpholinos revealed a robust role for the downstream target Chordin. Remarkably, Chordin depletion prevented all ectopic effects resulting from microinjection of the mRNA encoding the maternal cytoplasmic determinant Huluwa, including the radial expansion of brain tissue and the ectopic expression of the ventral gene sizzled. The main conclusion was that the BCNE and Nieuwkoop centers provide a double assurance mechanism for axial formation by independently activating similar downstream transcriptional target gene repertoires. We suggest that Siamois likely evolved from an ancestral Mix-type homeodomain protein called Sebox as a Xenopus-specific adaptation for the rapid differentiation of the anterior neural plate in the ectoderm.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA90095-1662
| | - Edward M. De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA90095-1662
| |
Collapse
|
2
|
Hongo I, Yamaguchi C, Okamoto H. Brain enlargement with rostral bias in larvae from a spontaneously occurring female variant line of Xenopus; role of aberrant embryonic Wnt/β-catenin signaling. Cells Dev 2024; 179:203918. [PMID: 38574816 DOI: 10.1016/j.cdev.2024.203918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
Increased brain size and its rostral bias are hallmarks of vertebrate evolution, but the underlying developmental and genetic basis remains poorly understood. To provide clues to understanding vertebrate brain evolution, we investigated the developmental mechanisms of brain enlargement observed in the offspring of a previously unrecognized, spontaneously occurring female variant line of Xenopus that appears to reflect a genetic variation. Brain enlargement in larvae from this line showed a pronounced rostral bias that could be traced back to the neural plate, the primordium of the brain. At the gastrula stage, the Spemann organizer, which is known to induce the neural plate from the adjacent dorsal ectoderm and give it the initial rostrocaudal patterning, was expanded from dorsal to ventral in a large proportion of the offspring of variant females. Consistently, siamois expression, which is required for Spemann organizer formation, was expanded laterally from dorsal to ventral at the blastula stage in variant offspring. This implies that the active region of the Wnt/β-catenin signaling pathway was similarly expanded in advance on the dorsal side, as siamois is a target gene of this pathway. Notably, the earliest detectable change in variant offspring was in fertilized eggs, in which maternal wnt11b mRNA, a candidate dorsalizing factor responsible for activating Wnt/β-catenin signaling in the dorsal embryonic region, had a wider distribution in the vegetal cortical cytoplasm. Since lateral spreading of wnt11b mRNA, and possibly that of other potential maternal dorsalizing factors in these eggs, is expected to facilitate lateral expansion of the active region of the Wnt/β-catenin pathway during subsequent embryonic stages, we concluded that aberrant Wnt/β-catenin signaling could cause rostral-biased brain enlargement via expansion of siamois expression and consequent expansion of the Spemann organizer in Xenopus. Our studies of spontaneously occurring variations in brain development in Xenopus would provide hints for uncovering genetic mutations that drive analogous morphogenetic variations during vertebrate brain evolution.
Collapse
Affiliation(s)
- Ikuko Hongo
- Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| | - Chihiro Yamaguchi
- Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| | - Harumasa Okamoto
- Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan.
| |
Collapse
|
3
|
Asashima M, Satou-Kobayashi Y. Spemann-Mangold organizer and mesoderm induction. Cells Dev 2024; 178:203903. [PMID: 38295873 DOI: 10.1016/j.cdev.2024.203903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
The discovery of the Spemann-Mangold organizer strongly influenced subsequent research on embryonic induction, with research aiming to elucidate the molecular characteristics of organizer activity being currently underway. Herein, we review the history of research on embryonic induction, and describe how the mechanisms of induction phenomena and developmental processes have been investigated. Classical experiments investigating the differentiation capacity and inductive activity of various embryonic regions were conducted by many researchers, and important theories of region-specific induction and the concept for chain of induction were proposed. The transition from experimental embryology to developmental biology has enabled us to understand the mechanisms of embryonic induction at the molecular level. Consequently, many inducing substances and molecules such as transcriptional factors and peptide growth factors involved in the organizer formation were identified. One of peptide growth factors, activin, acts as a mesoderm- and endoderm-inducing substance. Activin induces several tissues and organs from the undifferentiated cell mass of amphibian embryos in a concentration-dependent manner. We review the extent to which we can control in vitro organogenesis from undifferentiated cells, and discuss the application to stem cell-based regenerative medicine based on insights gained from animal experiments, such as in amphibians.
Collapse
Affiliation(s)
- Makoto Asashima
- Advanced Comprehensive Research Organization, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan.
| | - Yumeko Satou-Kobayashi
- Advanced Comprehensive Research Organization, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan
| |
Collapse
|
4
|
Keum BR, Yeo I, Koo Y, Han W, Choi SC, Kim GH, Han JK. Transmembrane protein 150b attenuates BMP signaling in the Xenopus organizer. J Cell Physiol 2023; 238:1850-1866. [PMID: 37435758 DOI: 10.1002/jcp.31059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 07/13/2023]
Abstract
The vertebrate organizer is a specified embryonic tissue that regulates dorsoventral patterning and axis formation. Although numerous cellular signaling pathways have been identified as regulators of the organizer's dynamic functions, the process remains incompletely understood, and as-yet unknown pathways remain to be explored for sophisticated mechanistic understanding of the vertebrate organizer. To identify new potential key factors of the organizer, we performed complementary DNA (cDNA) microarray screening using organizer-mimicking Xenopus laevis tissue. This analysis yielded a list of prospective organizer genes, and we determined the role of six-transmembrane domain containing transmembrane protein 150b (Tmem150b) in organizer function. Tmem150b was expressed in the organizer region and induced by Activin/Nodal signaling. In X. laevis, Tmem150b knockdown resulted in head defects and a shortened body axis. Moreover, Tmem150b negatively regulated bone morphogenetic protein (BMP) signaling, likely via physical interaction with activin receptor-like kinase 2 (ALK2). These findings demonstrated that Tmem150b functions as a novel membrane regulatory factor of BMP signaling with antagonistic effects, contributing to the understanding of regulatory molecular mechanisms of organizer axis function. Investigation of additional candidate genes identified in the cDNA microarray analysis could further delineate the genetic networks of the organizer during vertebrate embryogenesis.
Collapse
Affiliation(s)
- Byeong-Rak Keum
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
- Research Center for drug development, CYPHARMA, Daejeon, Korea
| | - Inchul Yeo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Youngmu Koo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Wonhee Han
- Department of Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Gun-Hwa Kim
- Research Center for drug development, CYPHARMA, Daejeon, Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Korea
| | - Jin-Kwan Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| |
Collapse
|
5
|
Foxh1/Nodal Defines Context-Specific Direct Maternal Wnt/β-Catenin Target Gene Regulation in Early Development. iScience 2020; 23:101314. [PMID: 32650116 PMCID: PMC7347983 DOI: 10.1016/j.isci.2020.101314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/20/2020] [Accepted: 06/20/2020] [Indexed: 12/19/2022] Open
Abstract
Although Wnt/β-catenin signaling is generally conserved and well understood, the regulatory mechanisms controlling context-specific direct Wnt target gene expression in development and disease are still unclear. The onset of zygotic gene transcription in early embryogenesis represents an ideal, accessible experimental system to investigate context-specific direct Wnt target gene regulation. We combine transcriptomics using RNA-seq with genome-wide β-catenin association using ChIP-seq to identify stage-specific direct Wnt target genes. We propose coherent feedforward regulation involving two distinct classes of direct maternal Wnt target genes, which differ both in expression and persistence of β-catenin association. We discover that genomic β-catenin association overlaps with Foxh1-associated regulatory sequences and demonstrate that direct maternal Wnt target gene expression requires Foxh1 function and Nodal/Tgfβ signaling. Our results support a new paradigm for direct Wnt target gene co-regulation with context-specific mechanisms that will inform future studies of embryonic development and more widely stem cell-mediated homeostasis and human disease. Combining RNA-seq and β-catenin ChIP-seq identifies direct Wnt target genes Two distinct classes of direct maternal Wnt/β-catenin target genes can be discerned We propose coherent feedforward regulation of gene expression of the second class Maternal Wnt target gene expression of both classes requires Nodal/Foxh1 signaling
Collapse
|
6
|
Esmaeili M, Blythe SA, Tobias JW, Zhang K, Yang J, Klein PS. Chromatin accessibility and histone acetylation in the regulation of competence in early development. Dev Biol 2020; 462:20-35. [PMID: 32119833 PMCID: PMC7225061 DOI: 10.1016/j.ydbio.2020.02.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/29/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
As development proceeds, inductive cues are interpreted by competent tissues in a spatially and temporally restricted manner. While key inductive signaling pathways within competent cells are well-described at a molecular level, the mechanisms by which tissues lose responsiveness to inductive signals are not well understood. Localized activation of Wnt signaling before zygotic gene activation in Xenopus laevis leads to dorsal development, but competence to induce dorsal genes in response to Wnts is lost by the late blastula stage. We hypothesize that loss of competence is mediated by changes in histone modifications leading to a loss of chromatin accessibility at the promoters of Wnt target genes. We use ATAC-seq to evaluate genome-wide changes in chromatin accessibility across several developmental stages. Based on overlap with p300 binding, we identify thousands of putative cis-regulatory elements at the gastrula stage, including sites that lose accessibility by the end of gastrulation and are enriched for pluripotency factor binding motifs. Dorsal Wnt target gene promoters are not accessible after the loss of competence in the early gastrula while genes involved in mesoderm and neural crest development maintain accessibility at their promoters. Inhibition of histone deacetylases increases acetylation at the promoters of dorsal Wnt target genes and extends competence for dorsal gene induction by Wnt signaling. Histone deacetylase inhibition, however, is not sufficient to extend competence for mesoderm or neural crest induction. These data suggest that chromatin state regulates the loss of competence to inductive signals in a context-dependent manner.
Collapse
Affiliation(s)
- Melody Esmaeili
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shelby A Blythe
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - John W Tobias
- Penn Genomic Analysis Core and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Peter S Klein
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Departments of Medicine (Hematology-Oncology) and Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Xenopus SOX5 enhances myogenic transcription indirectly through transrepression. Dev Biol 2018; 442:262-275. [PMID: 30071218 DOI: 10.1016/j.ydbio.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/16/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
Abstract
In anamniotes, somite compartimentalization in the lateral somitic domain leads simultaneously to myotome and dermomyotome formation. In the myotome, Xenopus Sox5 is co-expressed with Myod1 in the course of myogenic differentiation. Here, we studied the function of Sox5 using a Myod1-induced myogenic transcription assay in pluripotent cells of animal caps. We found that Sox5 enhances myogenic transcription of muscle markers Des, Actc1, Ckm and MyhE3. The use of chimeric transactivating or transrepressive Sox5 proteins indicates that Sox5 acts as a transrepressor and indirectly stimulates myogenic transcription except for the slow muscle-specific genes Myh7L, Myh7S, Myl2 and Tnnc1. We showed that this role is shared by Sox6, which is structurally similar to Sox5, both belonging to the SoxD subfamily of transcription factors. Moreover, Sox5 can antagonize the inhibitory function of Meox2 on myogenic differentiation. Meox2 which is a dermomyotome marker, represses myogenic transcription in Myod-induced myogenic transcription assay and in Nodal5-induced mesoderm from animal cap assay. The inhibitory function of Meox2 and the pro-myogenic function of Sox5 were confirmed during Xenopus normal development by the use of translation-blocking oligomorpholinos and dexamethasone inducible chimeric Sox5 and Meox2 proteins. We have therefore identified a new function for SoxD proteins in muscle cells, which can indirectly enhance myogenic transcription through transrepression, in addition to the previously identified function as a direct repressor of slow muscle-specific genes.
Collapse
|
8
|
Embryonic regeneration by relocalization of the Spemann organizer during twinning in Xenopus. Proc Natl Acad Sci U S A 2018; 115:E4815-E4822. [PMID: 29686106 PMCID: PMC6003488 DOI: 10.1073/pnas.1802749115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The formation of identical twins from a single egg has fascinated developmental biologists for a very long time. Previous work had shown that Xenopus blastulae bisected along the dorsal-ventral (D-V) midline (i.e., the sagittal plane) could generate twins but at very low frequencies. Here, we have improved this method by using an eyelash knife and changing saline solutions, reaching frequencies of twinning of 50% or more. This allowed mechanistic analysis of the twinning process. We unexpectedly observed that the epidermis of the resulting twins was asymmetrically pigmented at the tailbud stage of regenerating tadpoles. This pigment was entirely of maternal (oocyte) origin. Bisecting the embryo generated a large wound, which closed from all directions within 60 minutes, bringing cells normally fated to become Spemann organizer in direct contact with predicted ventral-most cells. Lineage-tracing analyses at the four-cell stage showed that in regenerating embryos midline tissues originated from the dorsal half, while the epidermis was entirely of ventral origin. Labeling of D-V segments at the 16-cell stage showed that the more pigmented epidermis originated from the ventral-most cells, while the less-pigmented epidermis arose from the adjoining ventral segment. This suggested a displacement of the organizer by 90°. Studies with the marker Chordin and phospho-Smad1/5/8 showed that in half embryos a new D-V gradient is intercalated at the site of the missing half. The displacement of self-organizing morphogen gradients uncovered here may help us understand not only twin formation in amphibians, but also rare cases of polyembryony.
Collapse
|
9
|
Teegala S, Chauhan R, Lei E, Weinstein DC. Tbx2 is required for the suppression of mesendoderm during early Xenopus development. Dev Dyn 2018; 247:903-913. [PMID: 29633424 DOI: 10.1002/dvdy.24633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/14/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND T-box family proteins are DNA-binding transcriptional regulators that play crucial roles during germ layer formation in the early vertebrate embryo. Well-characterized members of this family, including the transcriptional activators Brachyury and VegT, are essential for the proper formation of mesoderm and endoderm, respectively. To date, T-box proteins have not been shown to play a role in the promotion of the third primary germ layer, ectoderm. RESULTS Here, we report that the T-box factor Tbx2 is both sufficient and necessary for ectodermal differentiation in the frog Xenopus laevis. Tbx2 is expressed zygotically in the presumptive ectoderm, during blastula and gastrula stages. Ectopic expression of Tbx2 represses mesoderm and endoderm, while loss of Tbx2 leads to inappropriate expression of mesoderm- and endoderm-specific genes in the region fated to give rise to ectoderm. Misexpression of Tbx2 also promotes neural tissue in animal cap explants, suggesting that Tbx2 plays a role in both the establishment of ectodermal fate and its dorsoventral patterning. CONCLUSIONS Our studies demonstrate that Tbx2 functions as a transcriptional repressor during germ layer formation, and suggest that this activity is mediated in part through repression of target genes that are stimulated, in the mesendoderm, by transactivating T-box proteins. Taken together, our results point to a critical role for Tbx2 in limiting the potency of blastula-stage progenitor cells during vertebrate germ layer differentiation. Developmental Dynamics 247:903-913, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sushma Teegala
- Department of Biology, The Graduate Center, City University of New York, New York.,Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Riddhi Chauhan
- Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Emily Lei
- Department of Biology, Queens College, City University of New York, Flushing, New York
| | - Daniel C Weinstein
- Department of Biology, Queens College, City University of New York, Flushing, New York
| |
Collapse
|
10
|
Jin Y, Weinstein DC. Pitx1 regulates cement gland development in Xenopus laevis through activation of transcriptional targets and inhibition of BMP signaling. Dev Biol 2018. [PMID: 29530451 DOI: 10.1016/j.ydbio.2018.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The cement gland in Xenopus laevis has long been used as a model to study the interplay of cell signaling and transcription factors during embryogenesis. It has been shown that an intermediate level of Bone Morphogenetic Protein (BMP) signaling is essential for cement gland formation. In addition, several transcription factors have been linked to cement gland development. One of these, the homeodomain-containing protein Pitx1, can generate ectopic cement gland formation; however, the mechanisms underlying this process remain obscure. We report here, for the first time, a requirement for Pitx proteins in cement gland formation, in vivo: knockdown of both pitx1 and the closely related pitx2c inhibit endogenous cement gland formation. Pitx1 transcriptionally activates cement gland differentiation genes through both direct and indirect mechanisms, and functions as a transcriptional activator to inhibit BMP signaling. This inhibition, required for the expression of pitx genes, is partially mediated by Pitx1-dependent follistatin expression. Complete suppression of BMP signaling inhibits induction of cement gland markers by Pitx1; furthermore, we find that Pitx1 physically interacts with Smad1, an intracellular transducer of BMP signaling. We propose a model of cement gland formation in which Pitx1 limits local BMP signaling within the cement gland primordium, and recruits Smad1 to activate direct downstream targets.
Collapse
Affiliation(s)
- Ye Jin
- Program in Biology, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Daniel C Weinstein
- Department of Biology, Queens College, The City University of New York, 65-30 Kissena Boulevard, Queens, NY 11367, USA.
| |
Collapse
|
11
|
Bohnenpoll T, Wittern AB, Mamo TM, Weiss AC, Rudat C, Kleppa MJ, Schuster-Gossler K, Wojahn I, Lüdtke THW, Trowe MO, Kispert A. A SHH-FOXF1-BMP4 signaling axis regulating growth and differentiation of epithelial and mesenchymal tissues in ureter development. PLoS Genet 2017; 13:e1006951. [PMID: 28797033 PMCID: PMC5567910 DOI: 10.1371/journal.pgen.1006951] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 08/22/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022] Open
Abstract
The differentiated cell types of the epithelial and mesenchymal tissue compartments of the mature ureter of the mouse arise in a precise temporal and spatial sequence from uncommitted precursor cells of the distal ureteric bud epithelium and its surrounding mesenchyme. Previous genetic efforts identified a member of the Hedgehog (HH) family of secreted proteins, Sonic hedgehog (SHH) as a crucial epithelial signal for growth and differentiation of the ureteric mesenchyme. Here, we used conditional loss- and gain-of-function experiments of the unique HH signal transducer Smoothened (SMO) to further characterize the cellular functions and unravel the effector genes of HH signaling in ureter development. We showed that HH signaling is not only required for proliferation and SMC differentiation of cells of the inner mesenchymal region but also for survival of cells of the outer mesenchymal region, and for epithelial proliferation and differentiation. We identified the Forkhead transcription factor gene Foxf1 as a target of HH signaling in the ureteric mesenchyme. Expression of a repressor version of FOXF1 in this tissue completely recapitulated the mesenchymal and epithelial proliferation and differentiation defects associated with loss of HH signaling while re-expression of a wildtype version of FOXF1 in the inner mesenchymal layer restored these cellular programs when HH signaling was inhibited. We further showed that expression of Bmp4 in the ureteric mesenchyme depends on HH signaling and Foxf1, and that exogenous BMP4 rescued cell proliferation and epithelial differentiation in ureters with abrogated HH signaling or FOXF1 function. We conclude that SHH uses a FOXF1-BMP4 module to coordinate the cellular programs for ureter elongation and differentiation, and suggest that deregulation of this signaling axis occurs in human congenital anomalies of the kidney and urinary tract (CAKUT). The mammalian ureter is a simple tube with a specialized multi-layered epithelium, the urothelium, and a surrounding coat of fibroblasts and peristaltically active smooth muscle cells. Besides its important function in urinary drainage, the ureter represents a simple model system to study epithelial and mesenchymal tissue interactions in organ development. The differentiated cell types of the ureter coordinately arise from precursor cells of the distal ureteric bud and its surrounding mesenchyme. How their survival, growth and differentiation is regulated and coordinated within and between the epithelial and mesenchymal tissue compartments is largely unknown. Previous work identified Sonic hedgehog (SHH) as a crucial epithelial signal for growth and differentiation of the ureteric mesenchyme, but the entirety of the cellular functions and the molecular mediators of its mesenchymal signaling pathway have remained obscure. Here we showed that epithelial SHH acts in a paracrine fashion onto the ureteric mesenchyme to activate a FOXF1-BMP4 regulatory module that directs growth and differentiation of both ureteric tissue compartments. HH signaling additionally acts in outer mesenchymal cells as a survival factor. Thus, SHH is an epithelial signal that coordinates various cellular programs in early ureter development.
Collapse
Affiliation(s)
- Tobias Bohnenpoll
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Anna B. Wittern
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tamrat M. Mamo
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Carsten Rudat
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Marc-Jens Kleppa
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | - Irina Wojahn
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Timo H.-W. Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
12
|
Identification and comparative analyses of Siamois cluster genes in Xenopus laevis and tropicalis. Dev Biol 2017; 426:374-383. [PMID: 27522305 DOI: 10.1016/j.ydbio.2016.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/13/2016] [Accepted: 07/16/2016] [Indexed: 11/21/2022]
Abstract
Two siamois-related homeobox genes siamois (sia1) and twin (sia2), have been reported in Xenopus laevis. These genes are expressed in the blastula chordin- and noggin-expressing (BCNE) center and the Nieuwkoop center, and have complete secondary axis-inducing activity when over-expressed on the ventral side of the embryo. Using whole genome sequences of X. tropicalis and X. laevis, we identified two additional siamois-related genes, which are tandemly duplicated near sia1 and sia2 to form the siamois gene cluster. Four siamois genes in X. tropicalis are transcribed at blastula to gastrula stages. In X. laevis, the siamois gene cluster is present on both homeologous chromosomes, XLA3L and XLA3S. Transcripts from seven siamois genes (three on XLA3L and four on XLA3S) in X. laevis were detected at blastula to gastrula stages. A transcribed gene, sia1p. S, encodes an inactive protein without a homeodomain. When over-expressed ventrally, all siamois-related genes tested in this study except for sia1p. S induced a complete secondary axis, indicating that X. tropicalis and X. laevis have four and six active siamois-related genes, respectively. Of note, each gene required different amounts of mRNA for full activity. These results suggest the possibility that siamois cluster genes have functional redundancy to endow robustness and quickness to organizer formation in Xenopus species.
Collapse
|
13
|
Spemann organizer transcriptome induction by early beta-catenin, Wnt, Nodal, and Siamois signals in Xenopus laevis. Proc Natl Acad Sci U S A 2017; 114:E3081-E3090. [PMID: 28348214 DOI: 10.1073/pnas.1700766114] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The earliest event in Xenopus development is the dorsal accumulation of nuclear β-catenin under the influence of cytoplasmic determinants displaced by fertilization. In this study, a genome-wide approach was used to examine transcription of the 43,673 genes annotated in the Xenopus laevis genome under a variety of conditions that inhibit or promote formation of the Spemann organizer signaling center. Loss of function of β-catenin with antisense morpholinos reproducibly reduced the expression of 247 mRNAs at gastrula stage. Interestingly, only 123 β-catenin targets were enriched on the dorsal side and defined an early dorsal β-catenin gene signature. These genes included several previously unrecognized Spemann organizer components. Surprisingly, only 3 of these 123 genes overlapped with the late Wnt signature recently defined by two other groups using inhibition by Dkk1 mRNA or Wnt8 morpholinos, which indicates that the effects of β-catenin/Wnt signaling in early development are exquisitely regulated by stage-dependent mechanisms. We analyzed transcriptome responses to a number of treatments in a total of 46 RNA-seq libraries. These treatments included, in addition to β-catenin depletion, regenerating dorsal and ventral half-embryos, lithium chloride treatment, and the overexpression of Wnt8, Siamois, and Cerberus mRNAs. Only some of the early dorsal β-catenin signature genes were activated at blastula whereas others required the induction of endomesoderm, as indicated by their inhibition by Cerberus overexpression. These comprehensive data provide a rich resource for analyzing how the dorsal and ventral regions of the embryo communicate with each other in a self-organizing vertebrate model embryo.
Collapse
|
14
|
Charney RM, Paraiso KD, Blitz IL, Cho KWY. A gene regulatory program controlling early Xenopus mesendoderm formation: Network conservation and motifs. Semin Cell Dev Biol 2017; 66:12-24. [PMID: 28341363 DOI: 10.1016/j.semcdb.2017.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/12/2017] [Accepted: 03/20/2017] [Indexed: 02/08/2023]
Abstract
Germ layer formation is among the earliest differentiation events in metazoan embryos. In triploblasts, three germ layers are formed, among which the endoderm gives rise to the epithelial lining of the gut tube and associated organs including the liver, pancreas and lungs. In frogs (Xenopus), where early germ layer formation has been studied extensively, the process of endoderm specification involves the interplay of dozens of transcription factors. Here, we review the interactions between these factors, summarized in a transcriptional gene regulatory network (GRN). We highlight regulatory connections conserved between frog, fish, mouse, and human endodermal lineages. Especially prominent is the conserved role and regulatory targets of the Nodal signaling pathway and the T-box transcription factors, Vegt and Eomes. Additionally, we highlight network topologies and motifs, and speculate on their possible roles in development.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
15
|
Park DS, Yoon GH, Lee HS, Choi SC. Capsaicin inhibits the Wnt/β-catenin signaling pathway by down-regulating PP2A. Biochem Biophys Res Commun 2016; 478:455-461. [DOI: 10.1016/j.bbrc.2016.06.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/15/2016] [Indexed: 02/01/2023]
|
16
|
Gillis WQ, Kirmizitas A, Iwasaki Y, Ki DH, Wyrick JM, Thomsen GH. Gtpbp2 is a positive regulator of Wnt signaling and maintains low levels of the Wnt negative regulator Axin. Cell Commun Signal 2016; 14:15. [PMID: 27484226 PMCID: PMC4969687 DOI: 10.1186/s12964-016-0138-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/29/2016] [Indexed: 12/29/2022] Open
Abstract
Background Canonical Wnt signals, transduced by stabilized β-catenin, play similar roles across animals in maintaining stem cell pluripotency, regulating cell differentiation, and instructing normal embryonic development. Dysregulated Wnt/β-catenin signaling causes diseases and birth defects, and a variety of regulatory processes control this pathway to ensure its proper function and integration with other signaling systems. We previously identified GTP-binding protein 2 (Gtpbp2) as a novel regulator of BMP signaling, however further exploration revealed that Gtpbp2 can also affect Wnt signaling, which is a novel finding reported here. Results Knockdown of Gtpbp2 in Xenopus embryos causes severe axial defects and reduces expression of Spemann-Mangold organizer genes. Gtpbp2 knockdown blocks responses to ectopic Wnt8 ligand, such as organizer gene induction in ectodermal tissue explants and induction of secondary axes in whole embryos. However, organizer gene induction by ectopic Nodal2 is unaffected by Gtpbp2 knockdown. Epistasis tests, conducted by activating Wnt signal transduction at sequential points in the canonical pathway, demonstrate that Gtpbp2 is required downstream of Dishevelled and Gsk3β but upstream of β-catenin, which is similar to the previously reported effects of Axin1 overexpression in Xenopus embryos. Focusing on Axin in Xenopus embryos, we find that knockdown of Gtpbp2 elevates endogenous or exogenous Axin protein levels. Furthermore, Gtpbp2 fusion proteins co-localize with Dishevelled and co-immunoprecipitate with Axin and Gsk3b. Conclusions We conclude that Gtpbp2 is required for canonical Wnt/β-catenin signaling in Xenopus embryos. Our data suggest a model in which Gtpbp2 suppresses the accumulation of Axin protein, a rate-limiting component of the β-catenin destruction complex, such that Axin protein levels negatively correlate with Gtpbp2 levels. This model is supported by the similarity of our Gtpbp2-Wnt epistasis results and previously reported effects of Axin overexpression, the physical interactions of Gtpbp2 with Axin, and the correlation between elevated Axin protein levels and lost Wnt responsiveness upon Gtpbp2 knockdown. A wide variety of cancer-causing Wnt pathway mutations require low Axin levels, so development of Gtpbp2 inhibitors may provide a new therapeutic strategy to elevate Axin and suppress aberrant β-catenin signaling in cancer and other Wnt-related diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0138-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William Q Gillis
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA.,Present Address: Department of Biological Sciences, State University of New York, College at Old Westbury, Old Westbury, NY, 11568, USA
| | - Arif Kirmizitas
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA.,Present Address: The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Yasuno Iwasaki
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA
| | - Dong-Hyuk Ki
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA.,Present Address: Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Jonathan M Wyrick
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA
| | - Gerald H Thomsen
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, 11794-5215, USA.
| |
Collapse
|
17
|
Gaur S, Mandelbaum M, Herold M, Majumdar HD, Neilson KM, Maynard TM, Mood K, Daar IO, Moody SA. Neural transcription factors bias cleavage stage blastomeres to give rise to neural ectoderm. Genesis 2016; 54:334-49. [PMID: 27092474 PMCID: PMC4912902 DOI: 10.1002/dvg.22943] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/13/2016] [Accepted: 04/15/2016] [Indexed: 01/23/2023]
Abstract
The decision by embryonic ectoderm to give rise to epidermal versus neural derivatives is the result of signaling events during blastula and gastrula stages. However, there also is evidence in Xenopus that cleavage stage blastomeres contain maternally derived molecules that bias them toward a neural fate. We used a blastomere explant culture assay to test whether maternally deposited transcription factors bias 16-cell blastomere precursors of epidermal or neural ectoderm to express early zygotic neural genes in the absence of gastrulation interactions or exogenously supplied signaling factors. We found that Foxd4l1, Zic2, Gmnn, and Sox11 each induced explants made from ventral, epidermis-producing blastomeres to express early neural genes, and that at least some of the Foxd4l1 and Zic2 activities are required at cleavage stages. Similarly, providing extra Foxd4l1 or Zic2 to explants made from dorsal, neural plate-producing blastomeres significantly increased the expression of early neural genes, whereas knocking down either significantly reduced them. These results show that maternally delivered transcription factors bias cleavage stage blastomeres to a neural fate. We demonstrate that mouse and human homologs of Foxd4l1 have similar functional domains compared to the frog protein, as well as conserved transcriptional activities when expressed in Xenopus embryos and blastomere explants. genesis 54:334-349, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shailly Gaur
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
| | - Max Mandelbaum
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
| | - Mona Herold
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
| | - Himani Datta Majumdar
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
| | - Karen M. Neilson
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
| | | | - Kathy Mood
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Ira O. Daar
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Sally A. Moody
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, NW, Washington DC, USA
- George Washington University Institute for Neuroscience
| |
Collapse
|
18
|
Klein SL, Moody SA. Early neural ectodermal genes are activated by Siamois and Twin during blastula stages. Genesis 2015; 53:308-20. [PMID: 25892704 PMCID: PMC8943805 DOI: 10.1002/dvg.22854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 12/13/2022]
Abstract
BMP signaling distinguishes between neural and non-neural fates by activating epidermis-specific transcription and repressing neural-specific transcription. The neural ectoderm forms after the Organizer secrets antagonists that prevent these BMP-mediated activities. However, it is not known whether neural genes also are transcriptionally activated. Therefore, we tested the ability of nine Organizer transcription factors to ectopically induce the expression of four neural ectodermal genes in epidermal precursors. We found evidence for two pathways: Foxd4 and Sox11 were only induced by Sia and Twn, whereas Gmnn and Zic2 were induced by Sia, Twn, as well as seven other Organizer transcription factors. The induction of Foxd4, Gmnn and Zic2 by Sia/Twn was both non-cell autonomous (requiring an intermediate protein) and cell autonomous (direct), whereas the induction of Sox11 required Foxd4 activity. Because direct induction by Sia/Twn could occur endogenously in the dorsal-equatorial blastula cells that give rise to both the Organizer mesoderm and the neural ectoderm, we knocked down Sia/Twn in those cells. This prevented the blastula expression of Foxd4 and Sox11, demonstrating that Sia/Twn directly activate some neural genes before the separation of the Organizer mesoderm and neural ectoderm lineages.
Collapse
Affiliation(s)
- Steven L. Klein
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, Northwest, Washington, DC
| | - Sally A. Moody
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, 2300 I Street, Northwest, Washington, DC
| |
Collapse
|
19
|
Swift MR, Pham VN, Castranova D, Bell K, Poole RJ, Weinstein BM. SoxF factors and Notch regulate nr2f2 gene expression during venous differentiation in zebrafish. Dev Biol 2014; 390:116-25. [PMID: 24699544 DOI: 10.1016/j.ydbio.2014.03.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/03/2014] [Accepted: 03/25/2014] [Indexed: 11/19/2022]
Abstract
Initial embryonic determination of artery or vein identity is regulated by genetic factors that work in concert to specify the endothelial cell׳s (EC) fate, giving rise to two structurally unique components of the circulatory loop. The Shh/VEGF/Notch pathway is critical for arterial specification, while the orphan receptor nr2f2 (COUP-TFII) has been implicated in venous specification. Studies in mice have shown that nr2f2 is expressed in venous but not arterial ECs, and that it preferentially induces markers of venous cell fate. We have examined the role of nr2f2 during early arterial-venous development in the zebrafish trunk. We show that expression of a subset of markers of venous endothelial identity requires nr2f2, while the expression of nr2f2 itself requires sox7 and sox18 gene function. However, while sox7 and sox18 are expressed in both the cardinal vein and the dorsal aorta during early trunk development, nr2f2 is expressed only in the cardinal vein. We show that Notch signaling activity present in the dorsal aorta suppresses expression of nr2f2, restricting nr2f2-dependent promotion of venous differentiation to the cardinal vein.
Collapse
Affiliation(s)
- Matthew R Swift
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kameha Bell
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard J Poole
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Brant M Weinstein
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Vandenberg LN, Levin M. A unified model for left-right asymmetry? Comparison and synthesis of molecular models of embryonic laterality. Dev Biol 2013; 379:1-15. [PMID: 23583583 PMCID: PMC3698617 DOI: 10.1016/j.ydbio.2013.03.021] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/15/2013] [Accepted: 03/22/2013] [Indexed: 12/31/2022]
Abstract
Understanding how and when the left-right (LR) axis is first established is a fundamental question in developmental biology. A popular model is that the LR axis is established relatively late in embryogenesis, due to the movement of motile cilia and the resultant directed fluid flow during late gastrulation/early neurulation. Yet, a large body of evidence suggests that biophysical, molecular, and bioelectrical asymmetries exist much earlier in development, some as early as the first cell cleavage after fertilization. Alternative models of LR asymmetry have been proposed that accommodate these data, postulating that asymmetry is established due to a chiral cytoskeleton and/or the asymmetric segregation of chromatids. There are some similarities, and many differences, in how these various models postulate the origin and timing of symmetry breaking and amplification, and these events' linkage to the well-conserved subsequent asymmetric transcriptional cascades. This review examines experimental data that lend strong support to an early origin of LR asymmetry, yet are also consistent with later roles for cilia in the amplification of LR pathways. In this way, we propose that the various models of asymmetry can be unified: early events are needed to initiate LR asymmetry, and later events could be utilized by some species to maintain LR-biases. We also present an alternative hypothesis, which proposes that individual embryos stochastically choose one of several possible pathways with which to establish their LR axis. These two hypotheses are both tractable in appropriate model species; testing them to resolve open questions in the field of LR patterning will reveal interesting new biology of wide relevance to developmental, cell, and evolutionary biology.
Collapse
Affiliation(s)
- Laura N. Vandenberg
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University, Medford, MA 02155
| | - Michael Levin
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University, Medford, MA 02155
| |
Collapse
|
21
|
Abstract
The Wnt pathway is a major embryonic signaling pathway that controls cell proliferation, cell fate, and body-axis determination in vertebrate embryos. Soon after egg fertilization, Wnt pathway components play a role in microtubule-dependent dorsoventral axis specification. Later in embryogenesis, another conserved function of the pathway is to specify the anteroposterior axis. The dual role of Wnt signaling in Xenopus and zebrafish embryos is regulated at different developmental stages by distinct sets of Wnt target genes. This review highlights recent progress in the discrimination of different signaling branches and the identification of specific pathway targets during vertebrate axial development.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | | |
Collapse
|
22
|
Landin Malt A, Cagliero J, Legent K, Silber J, Zider A, Flagiello D. Alteration of TEAD1 expression levels confers apoptotic resistance through the transcriptional up-regulation of Livin. PLoS One 2012; 7:e45498. [PMID: 23029054 PMCID: PMC3454436 DOI: 10.1371/journal.pone.0045498] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/17/2012] [Indexed: 11/19/2022] Open
Abstract
Background TEA domain (TEAD) proteins are highly conserved transcription factors involved in embryonic development and differentiation of various tissues. More recently, emerging evidences for a contribution of these proteins towards apoptosis and cell proliferation regulation have also been proposed. These effects appear to be mediated by the interaction between TEAD and its co-activator Yes-Associated Protein (YAP), the downstream effector of the Hippo tumour suppressor pathway. Methodology/Principal Findings We further investigated the mechanisms underlying TEAD-mediated apoptosis regulation and showed that overexpression or RNAi-mediated silencing of the TEAD1 protein is sufficient to protect mammalian cell lines from induced apoptosis, suggesting a proapoptotic function for TEAD1 and a non physiological cytoprotective effect for overexpressed TEAD1. Moreover we show that the apoptotic resistance conferred by altered TEAD1 expression is mediated by the transcriptional up-regulation of Livin, a member of the Inhibitor of Apoptosis Protein (IAP) family. In addition, we show that overexpression of a repressive form of TEAD1 can induce Livin up-regulation, indicating that the effect of TEAD1 on Livin expression is indirect and favoring a model in which TEAD1 activates a repressor of Livin by interacting with a limiting cofactor that gets titrated upon TEAD1 up-regulation. Interestingly, we show that overexpression of a mutated form of TEAD1 (Y421H) implicated in Sveinsson's chorioretinal atrophy that strongly reduces its interaction with YAP as well as its activation, can induce Livin expression and protect cells from induced apoptosis, suggesting that YAP is not the cofactor involved in this process. Conclusions/Significance Taken together our data reveal a new, Livin-dependent, apoptotic role for TEAD1 in mammals and provide mechanistic insight downstream of TEAD1 deregulation in cancers.
Collapse
Affiliation(s)
| | | | | | | | - Alain Zider
- Univ Paris Diderot, Sorbonne Paris Cité, Equipe de Génétique Moléculaire de la Différenciation, IJM, UMR 7592 CNRS, Paris, France
- * E-mail: (AZ); (DF)
| | - Domenico Flagiello
- Univ Paris Diderot, Sorbonne Paris Cité, Equipe de Génétique Moléculaire de la Différenciation, IJM, UMR 7592 CNRS, Paris, France
- * E-mail: (AZ); (DF)
| |
Collapse
|
23
|
Robertshaw E, Kiecker C. Phylogenetic origins of brain organisers. SCIENTIFICA 2012; 2012:475017. [PMID: 24278699 PMCID: PMC3820451 DOI: 10.6064/2012/475017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 06/21/2012] [Indexed: 06/02/2023]
Abstract
The regionalisation of the nervous system begins early in embryogenesis, concomitant with the establishment of the anteroposterior (AP) and dorsoventral (DV) body axes. The molecular mechanisms that drive axis induction appear to be conserved throughout the animal kingdom and may be phylogenetically older than the emergence of bilateral symmetry. As a result of this process, groups of patterning genes that are equally well conserved are expressed at specific AP and DV coordinates of the embryo. In the emerging nervous system of vertebrate embryos, this initial pattern is refined by local signalling centres, secondary organisers, that regulate patterning, proliferation, and axonal pathfinding in adjacent neuroepithelium. The main secondary organisers for the AP neuraxis are the midbrain-hindbrain boundary, zona limitans intrathalamica, and anterior neural ridge and for the DV neuraxis the notochord, floor plate, and roof plate. A search for homologous secondary organisers in nonvertebrate lineages has led to controversy over their phylogenetic origins. Based on a recent study in hemichordates, it has been suggested that the AP secondary organisers evolved at the base of the deuterostome superphylum, earlier than previously thought. According to this view, the lack of signalling centres in some deuterostome lineages is likely to reflect a secondary loss due to adaptive processes. We propose that the relative evolutionary flexibility of secondary organisers has contributed to a broader morphological complexity of nervous systems in different clades.
Collapse
Affiliation(s)
- Ellen Robertshaw
- MRC Centre for Developmental Neurobiology, King's College London, 4th Floor, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| | - Clemens Kiecker
- MRC Centre for Developmental Neurobiology, King's College London, 4th Floor, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| |
Collapse
|
24
|
Reid CD, Zhang Y, Sheets MD, Kessler DS. Transcriptional integration of Wnt and Nodal pathways in establishment of the Spemann organizer. Dev Biol 2012; 368:231-41. [PMID: 22627292 DOI: 10.1016/j.ydbio.2012.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/22/2012] [Accepted: 05/08/2012] [Indexed: 11/25/2022]
Abstract
Signaling inputs from multiple pathways are essential for the establishment of distinct cell and tissue types in the embryo. Therefore, multiple signals must be integrated to activate gene expression and confer cell fate, but little is known about how this occurs at the level of target gene promoters. During early embryogenesis, Wnt and Nodal signals are required for formation of the Spemann organizer, which is essential for germ layer patterning and axis formation. Signaling by both Wnt and Nodal pathways is required for the expression of multiple organizer genes, suggesting that integration of these signals is required for organizer formation. Here, we demonstrate transcriptional cooperation between the Wnt and Nodal pathways in the activation of the organizer genes Goosecoid (Gsc), Cerberus (Cer), and Chordin (Chd). Combined Wnt and Nodal signaling synergistically activates transcription of these organizer genes. Effectors of both pathways occupy the Gsc, Cer and Chd promoters and effector occupancy is enhanced with active Wnt and Nodal signaling. This suggests that, at organizer gene promoters, a stable transcriptional complex containing effectors of both pathways forms in response to combined Wnt and Nodal signaling. Consistent with this idea, the histone acetyltransferase p300 is recruited to organizer promoters in a Wnt and Nodal effector-dependent manner. Taken together, these results offer a mechanism for spatial and temporal restriction of organizer gene transcription by the integration of two major signaling pathways, thus establishing the Spemann organizer domain.
Collapse
Affiliation(s)
- Christine D Reid
- Department of Cell and Developmental Biology, University of Pennsylvania, School of Medicine, Room 1110 Biomedical Research Building 2/3, 421 Curie Boulevard, Philadelphia, PA 19104-6058, USA
| | | | | | | |
Collapse
|
25
|
Xmab21l3 mediates dorsoventral patterning in Xenopus laevis. Mech Dev 2012; 129:136-46. [PMID: 22609272 DOI: 10.1016/j.mod.2012.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 02/05/2023]
Abstract
Specification of the dorsoventral (DV) axis is critical for the subsequent differentiation of regional fate in the primary germ layers of the vertebrate embryo. We have identified a novel factor that is essential for dorsal development in embryos of the frog Xenopus laevis. Misexpression of Xenopus mab21-like 3 (Xmab21l3) dorsalizes gastrula-stage mesoderm and neurula-stage ectoderm, while morpholino-mediated knockdown of Xmab21l3 inhibits dorsal differentiation of these embryonic germ layers. Xmab21l3 is a member of a chordate-specific subclass of a recently characterized gene family, all members of which contain a conserved, but as yet ill-defined, Mab21 domain. Our studies suggest that Xmab21l3 functions to repress ventralizing activity in the early vertebrate embryo, via regulation of BMP/Smad and Ras/ERK signaling.
Collapse
|
26
|
Xenopus Zic3 controls notochord and organizer development through suppression of the Wnt/β-catenin signaling pathway. Dev Biol 2012; 361:220-31. [DOI: 10.1016/j.ydbio.2011.10.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 09/30/2011] [Accepted: 10/08/2011] [Indexed: 11/21/2022]
|
27
|
Douville JM, Cheung DYC, Herbert KL, Moffatt T, Wigle JT. Mechanisms of MEOX1 and MEOX2 regulation of the cyclin dependent kinase inhibitors p21 and p16 in vascular endothelial cells. PLoS One 2011; 6:e29099. [PMID: 22206000 PMCID: PMC3243699 DOI: 10.1371/journal.pone.0029099] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 11/21/2011] [Indexed: 12/31/2022] Open
Abstract
Senescence, the state of permanent cell cycle arrest, has been associated
with endothelial cell dysfunction and atherosclerosis. The cyclin dependent
kinase inhibitors p21CIP1/WAF1 and p16INK4a govern the
G1/S cell cycle checkpoint and are essential for determining whether
a cell enters into an arrested state. The homeodomain transcription factor
MEOX2 is an important regulator of vascular cell proliferation and is a direct
transcriptional activator of both p21CIP1/WAF1 and p16INK4a.
MEOX1 and MEOX2 have been shown to be partially functionally redundant during
development, suggesting that they regulate similar target genes in
vivo. We compared the ability of MEOX1 and MEOX2 to activate p21CIP1/WAF1
and p16INK4a expression and induce endothelial cell cycle arrest.
Our results demonstrate for the first time that MEOX1 regulates the MEOX2
target genes p21CIP1/WAF1 and p16INK4a. In addition,
increased expression of either of the MEOX homeodomain transcription factors
leads to cell cycle arrest and endothelial cell senescence. Furthermore, we
show that the mechanism of transcriptional activation of these cyclin dependent
kinase inhibitor genes by MEOX1 and MEOX2 is distinct. MEOX1 and MEOX2 activate
p16INK4a in a DNA binding dependent manner, whereas they induce
p21CIP1/WAF1 in a DNA binding independent manner.
Collapse
Affiliation(s)
- Josette M. Douville
- Institute of Cardiovascular Sciences,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical
Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Y. C. Cheung
- Institute of Cardiovascular Sciences,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
| | - Krista L. Herbert
- Institute of Cardiovascular Sciences,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
| | - Teri Moffatt
- Institute of Cardiovascular Sciences,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
| | - Jeffrey T. Wigle
- Institute of Cardiovascular Sciences,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical
Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
28
|
Carneiro K, Donnet C, Rejtar T, Karger BL, Barisone GA, Díaz E, Kortagere S, Lemire JM, Levin M. Histone deacetylase activity is necessary for left-right patterning during vertebrate development. BMC DEVELOPMENTAL BIOLOGY 2011; 11:29. [PMID: 21599922 PMCID: PMC3113753 DOI: 10.1186/1471-213x-11-29] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 05/20/2011] [Indexed: 01/23/2023]
Abstract
Background Consistent asymmetry of the left-right (LR) axis is a crucial aspect of vertebrate embryogenesis. Asymmetric gene expression of the TGFβ superfamily member Nodal related 1 (Nr1) in the left lateral mesoderm plate is a highly conserved step regulating the situs of the heart and viscera. In Xenopus, movement of maternal serotonin (5HT) through gap-junctional paths at cleavage stages dictates asymmetry upstream of Nr1. However, the mechanisms linking earlier biophysical asymmetries with this transcriptional control point are not known. Results To understand how an early physiological gradient is transduced into a late, stable pattern of Nr1 expression we investigated epigenetic regulation during LR patterning. Embryos injected with mRNA encoding a dominant-negative of Histone Deacetylase (HDAC) lacked Nr1 expression and exhibited randomized sidedness of the heart and viscera (heterotaxia) at stage 45. Timing analysis using pharmacological blockade of HDACs implicated cleavage stages as the active period. Inhibition during these early stages was correlated with an absence of Nr1 expression at stage 21, high levels of heterotaxia at stage 45, and the deposition of the epigenetic marker H3K4me2 on the Nr1 gene. To link the epigenetic machinery to the 5HT signaling pathway, we performed a high-throughput proteomic screen for novel cytoplasmic 5HT partners associated with the epigenetic machinery. The data identified the known HDAC partner protein Mad3 as a 5HT-binding regulator. While Mad3 overexpression led to an absence of Nr1 transcription and randomized the LR axis, a mutant form of Mad3 lacking 5HT binding sites was not able to induce heterotaxia, showing that Mad3's biological activity is dependent on 5HT binding. Conclusion HDAC activity is a new LR determinant controlling the epigenetic state of Nr1 from early developmental stages. The HDAC binding partner Mad3 may be a new serotonin-dependent regulator of asymmetry linking early physiological asymmetries to stable changes in gene expression during organogenesis.
Collapse
Affiliation(s)
- Katia Carneiro
- Department of Biology Center for Regenerative and Developmental Biology Tufts University, Medford, MA 02155 USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Siamois and Twin are redundant and essential in formation of the Spemann organizer. Dev Biol 2011; 352:367-81. [PMID: 21295564 DOI: 10.1016/j.ydbio.2011.01.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 01/11/2011] [Accepted: 01/25/2011] [Indexed: 02/03/2023]
Abstract
The Spemann organizer is an essential signaling center in Xenopus germ layer patterning and axis formation. Organizer formation occurs in dorsal blastomeres receiving both maternal Wnt and zygotic Nodal signals. In response to stabilized βcatenin, dorsal blastomeres express the closely related transcriptional activators, Siamois (Sia) and Twin (Twn), members of the paired homeobox family. Sia and Twn induce organizer formation and expression of organizer-specific genes, including Goosecoid (Gsc). In spite of the similarity of Sia and Twn sequence and expression pattern, it is unclear whether these factors function equivalently in promoter binding and subsequent transcriptional activation, or if Sia and Twn are required for all aspects of organizer function. Here we report that Sia and Twn activate Gsc transcription by directly binding to a conserved P3 site within the Wnt-responsive proximal element of the Gsc promoter. Sia and Twn form homodimers and heterodimers by direct homeodomain interaction and dimer forms are indistinguishable in both DNA-binding and activation functions. Sequential chromatin immunoprecipitation reveals that the endogenous Gsc promoter can be occupied by either Sia or Twn homodimers or Sia-Twn heterodimers. Knockdown of Sia and Twn together, but not individually, results in a failure of organizer gene expression and a disruption of axis formation, consistent with a redundant role for Sia and Twn in organizer formation. Furthermore, simultaneous knockdown of Sia and Twn blocks axis induction in response to ectopic Wnt signaling, demonstrating an essential role for Sia and Twn in mediating the transcriptional response to the maternal Wnt pathway. The results demonstrate the functional redundancy of Sia and Twn and their essential role in direct transcriptional responses necessary for Spemann organizer formation.
Collapse
|
30
|
Rankin SA, Kormish J, Kofron M, Jegga A, Zorn AM. A gene regulatory network controlling hhex transcription in the anterior endoderm of the organizer. Dev Biol 2011; 351:297-310. [PMID: 21215263 DOI: 10.1016/j.ydbio.2010.11.037] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 11/15/2010] [Accepted: 11/17/2010] [Indexed: 10/18/2022]
Abstract
The homeobox gene hhex is one of the earliest markers of the anterior endoderm, which gives rise to foregut organs such as the liver, ventral pancreas, thyroid, and lungs. The regulatory networks controlling hhex transcription are poorly understood. In an extensive cis-regulatory analysis of the Xenopus hhex promoter, we determined how the Nodal, Wnt, and BMP pathways and their downstream transcription factors regulate hhex expression in the gastrula organizer. We show that Nodal signaling, present throughout the endoderm, directly activates hhex transcription via FoxH1/Smad2 binding sites in the proximal -0.44 Kb promoter. This positive action of Nodal is suppressed in the ventral-posterior endoderm by Vent 1 and Vent2, homeodomain repressors that are induced by BMP signaling. Maternal Wnt/β-catenin on the dorsal side of the embryo cooperates with Nodal and indirectly activates hhex expression via the homeodomain activators Siamois and Twin. Siamois/Twin stimulate hhex transcription through two mechanisms: (1) they induce the expression of Otx2 and Lim1 and together Siamois, Twin, Otx2, and Lim1 appear to promote hhex transcription through homeobox sites in a Wnt-responsive element located between -0.65 to -0.55 Kb of the hhex promoter. (2) Siamois/Twin also induce the expression of the BMP-antagonists Chordin and Noggin, which are required to exclude Vents from the organizer allowing hhex transcription. This study reveals a complex network regulating anterior endoderm transcription in the early embryo.
Collapse
Affiliation(s)
- Scott A Rankin
- Division of Developmental Biology, Cincinnati Children's Research Foundation and Department of Pediatrics, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
31
|
Abstract
A transcriptional corepressor, Xenopus furry (Xfurry), is expressed in the chordamesodermal region and induces secondary dorsal axes when overexpressed on the ventral side of the embryo. The N-terminal furry domain functions as a repressor, and the C-terminal leucine zipper (LZ) motifs /coiled-coil structure, found only in vertebrate homologs, contributes to the nuclear localization. The engrailed repressor (enR)+LZ repressor construct, which has properties similar to Xfurry, induced several chordamesodermal genes. In contrast, an antisense morpholino oligonucleotide, Xfurry-MO, and the activating construct, herpes simplex virus protein (VP16)+LZ, had effects opposite those of Xfurry overexpression. Because blocking protein synthesis with cycloheximide superinduced several Xfurry transcriptional targets, and because expression of enR+LZ induced such genes under cycloheximide treatment, we analyzed the role of an Xfurry transcriptional target, microRNA miR-15. Cycloheximide reduced the expression of primary miR-15 (pri-miR-15), whereas miR-15 reduced the expression of genes superinduced by cycloheximide treatment. These results show that Xfurry regulates chordamesodermal genes by contributing to repression of pretranscriptional gene silencing by miR-15.
Collapse
|
32
|
Vandenberg LN, Levin M. Consistent left-right asymmetry cannot be established by late organizers in Xenopus unless the late organizer is a conjoined twin. Development 2010; 137:1095-105. [PMID: 20215347 PMCID: PMC2835325 DOI: 10.1242/dev.041798] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2010] [Indexed: 12/28/2022]
Abstract
How embryos consistently orient asymmetries of the left-right (LR) axis is an intriguing question, as no macroscopic environmental cues reliably distinguish left from right. Especially unclear are the events coordinating LR patterning with the establishment of the dorsoventral (DV) axes and midline determination in early embryos. In frog embryos, consistent physiological and molecular asymmetries manifest by the second cell cleavage; however, models based on extracellular fluid flow at the node predict correct de novo asymmetry orientation during neurulation. We addressed these issues in Xenopus embryos by manipulating the timing and location of dorsal organizer induction: the primary dorsal organizer was ablated by UV irradiation, and a new organizer was induced at various locations, either early, by mechanical rotation, or late, by injection of lithium chloride (at 32 cells) or of the transcription factor XSiamois (which functions after mid-blastula transition). These embryos were then analyzed for the position of three asymmetric organs. Whereas organizers rescued before cleavage properly oriented the LR axis 90% of the time, organizers induced in any position at any time after the 32-cell stage exhibited randomized laterality. Late organizers were unable to correctly orient the LR axis even when placed back in their endogenous location. Strikingly, conjoined twins produced by late induction of ectopic organizers did have normal asymmetry. These data reveal that although correct LR orientation must occur no later than early cleavage stages in singleton embryos, a novel instructive influence from an early organizer can impose normal asymmetry upon late organizers in the same cell field.
Collapse
Affiliation(s)
- Laura N. Vandenberg
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Center for Regenerative and Developmental Biology, and Biology Department, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
33
|
Induction and patterning of trunk and tail neural ectoderm by the homeobox gene eve1 in zebrafish embryos. Proc Natl Acad Sci U S A 2010; 107:3564-9. [PMID: 20142486 DOI: 10.1073/pnas.1000389107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In vertebrates, Evx homeodomain transcription factor-encoding genes are expressed in the posterior region during embryonic development, and overexpression experiments have revealed roles in tail development in fish and frogs. We analyzed the molecular mechanisms of posterior neural development and axis formation regulated by eve1. We show that eve1 is involved in establishing trunk and tail neural ectoderm by two independent mechanisms: First, eve1 posteriorizes neural ectoderm via induction of aldh1a2, which encodes an enzyme that synthesizes retinoic acid; second, eve1 is involved in neural induction in the posterior ectoderm by attenuating BMP expression. Further, eve1 can restore trunk neural tube formation in the organizer-deficient ichabod(-/-) mutant. We conclude that eve1 is crucial for the organization of the antero-posterior and dorso-ventral axis in the gastrula ectoderm and also has trunk- and tail-promoting activity.
Collapse
|
34
|
Blimp1-mediated repression of negative regulators is required for osteoclast differentiation. Proc Natl Acad Sci U S A 2010; 107:3117-22. [PMID: 20133620 DOI: 10.1073/pnas.0912779107] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Regulation of irreversible cell lineage commitment depends on a delicate balance between positive and negative regulators, which comprise a sophisticated network of transcription factors. Receptor activator of NF-kappaB ligand (RANKL) stimulates the differentiation of bone-resorbing osteoclasts through the induction of nuclear factor of activated T cells c1 (NFATc1), the essential transcription factor for osteoclastogenesis. Osteoclast-specific robust induction of NFATc1 is achieved through an autoamplification mechanism, in which NFATc1 is constantly activated by calcium signaling while the negative regulators of NFATc1 are suppressed. However, it has been unclear how such negative regulators are repressed during osteoclastogenesis. Here we show that B lymphocyte-induced maturation protein-1 (Blimp1; encoded by Prdm1), which is induced by RANKL through NFATc1 during osteoclastogenesis, functions as a transcriptional repressor of anti-osteoclastogenic genes such as Irf8 and Mafb. Overexpression of Blimp1 leads to an increase in osteoclast formation, and Prdm1-deficient osteoclast precursor cells do not undergo osteoclast differentiation efficiently. The importance of Blimp1 in bone homeostasis is underscored by the observation that mice with an osteoclast-specific deficiency in the Prdm1 gene exhibit a high bone mass phenotype caused by a decreased number of osteoclasts. Thus, NFATc1 choreographs the determination of cell fate in the osteoclast lineage by inducing the repression of negative regulators as well as through its effect on positive regulators.
Collapse
|
35
|
Dixon Fox M, Bruce AEE. Short- and long-range functions of Goosecoid in zebrafish axis formation are independent of Chordin, Noggin 1 and Follistatin-like 1b. Development 2009; 136:1675-85. [PMID: 19369398 DOI: 10.1242/dev.031161] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The organizer is essential for dorsal-ventral (DV) patterning in vertebrates. Goosecoid (Gsc), a transcriptional repressor found in the organizer, elicits partial secondary axes when expressed ventrally in Xenopus, similar to an organizer transplant. Although gsc is expressed in all vertebrate organizers examined, knockout studies in mouse suggested that it is not required for DV patterning. Moreover, experiments in Xenopus and zebrafish suggest a role in head formation, although a function in axial mesoderm formation is less clear. To clarify the role of Gsc in vertebrate development, we used gain- and loss-of-function approaches in zebrafish. Ventral injection of low doses of gsc produced incomplete secondary axes, which we propose results from short-range repression of BMP signaling. Higher gsc doses resulted in complete secondary axes and long-range signaling, correlating with repression of BMP and Wnt signals. In striking contrast to Xenopus, the BMP inhibitor Chordin (Chd) is not required for Gsc function. Gsc produced complete secondary axes in chd null mutant embryos and gsc-morpholino knockdown in chd mutants enhanced the mutant phenotype, suggesting that Gsc has Chd-independent functions in DV patterning. Even more striking was that Gsc elicited complete secondary axes in the absence of three secreted BMP antagonists, Chd, Follistatin-like 1b and Noggin 1, suggesting that Gsc functions in parallel with secreted BMP inhibitors. Our findings suggest that Gsc has dose dependent effects on axis induction and provide new insights into molecularly distinct short- and long-range signaling activities of the organizer.
Collapse
Affiliation(s)
- Monica Dixon Fox
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | | |
Collapse
|
36
|
Lyons JP, Miller RK, Zhou X, Weidinger G, Deroo T, Denayer T, Park JI, Ji H, Hong JY, Li A, Moon RT, Jones EA, Vleminckx K, Vize PD, McCrea PD. Requirement of Wnt/beta-catenin signaling in pronephric kidney development. Mech Dev 2008; 126:142-59. [PMID: 19100832 DOI: 10.1016/j.mod.2008.11.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 11/24/2008] [Indexed: 01/02/2023]
Abstract
The pronephric kidney controls water and electrolyte balance during early fish and amphibian embryogenesis. Many Wnt signaling components have been implicated in kidney development. Specifically, in Xenopus pronephric development as well as the murine metanephroi, the secreted glycoprotein Wnt-4 has been shown to be essential for renal tubule formation. Despite the importance of Wnt signals in kidney organogenesis, little is known of the definitive downstream signaling pathway(s) that mediate their effects. Here we report that inhibition of Wnt/beta-catenin signaling within the pronephric field of Xenopus results in significant losses to kidney epithelial tubulogenesis with little or no effect on adjoining axis or somite development. We find that the requirement for Wnt/beta-catenin signaling extends throughout the pronephric primordium and is essential for the development of proximal and distal tubules of the pronephros as well as for the development of the duct and glomus. Although less pronounced than effects upon later pronephric tubule differentiation, inhibition of the Wnt/beta-catenin pathway decreased expression of early pronephric mesenchymal markers indicating it is also needed in early pronephric patterning. We find that upstream inhibition of Wnt/beta-catenin signals in zebrafish likewise reduces pronephric epithelial tubulogenesis. We also find that exogenous activation of Wnt/beta-catenin signaling within the Xenopus pronephric field results in significant tubulogenic losses. Together, we propose Wnt/beta-catenin signaling is required for pronephric tubule, duct and glomus formation in Xenopus laevis, and this requirement is conserved in zebrafish pronephric tubule formation.
Collapse
Affiliation(s)
- Jon P Lyons
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1000, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Luu O, Nagel M, Wacker S, Lemaire P, Winklbauer R. Control of gastrula cell motility by the Goosecoid/Mix.1/ Siamois network: basic patterns and paradoxical effects. Dev Dyn 2008; 237:1307-20. [PMID: 18407556 DOI: 10.1002/dvdy.21522] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In the vegetal half of the Xenopus gastrula, cell populations differ with respect to migration on fibronectin substratum. We show that the paired-class homeodomain transcription factors Goosecoid (Gsc), Mix.1, and Siamois (Sia) are involved in the modulation of migration velocity and cell polarity. Mix.1 is expressed in the whole vegetal half and serves as a competence factor that is necessary, but not sufficient, for rapid cell migration and polarization. In the head mesoderm, Gsc and Sia are coexpressed with Mix.1, promoting rapid cell migration and polarization. Ectopic expression of Gsc and Sia in both vegetal and ventral regions often generates paradoxical effects; if a factor activates a certain motility trait in one region, it inhibits it in the other. Migration velocity and cell polarity are regulated independently. Fast and efficiently migrating multipolar cells and slow-moving polarized cells can be obtained by ectopic expression of these transcription factors in different combinations.
Collapse
Affiliation(s)
- Olivia Luu
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
38
|
Keren A, Keren-Politansky A, Bengal E. A p38 MAPK-CREB pathway functions to pattern mesoderm in Xenopus. Dev Biol 2008; 322:86-94. [PMID: 18675264 DOI: 10.1016/j.ydbio.2008.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 06/29/2008] [Accepted: 07/07/2008] [Indexed: 12/30/2022]
Abstract
Dorsal-ventral patterning is specified by signaling centers secreting antagonizing morphogens that form a signaling gradient. Yet, how morphogen gradient is translated intracellularly into fate decisions remains largely unknown. Here, we report that p38 MAPK and CREB function along the dorsal-ventral axis in mesoderm patterning. We find that the phosphorylated form of CREB (S133) is distributed in a gradient along the dorsal-ventral mesoderm axis and that the p38 MAPK pathway mediates the phosphorylation of CREB. Knockdown of CREB prevents chordin expression and mesoderm dorsalization by the Spemann organizer, whereas ectopic expression of activated CREB-VP16 chimera induces chordin expression and dorsalizes mesoderm. Expression of high levels of p38 activator, MKK6E or CREB-VP16 in embryos converts ventral mesoderm into a dorsal organizing center. p38 MAPK and CREB function downstream of maternal Wnt/beta-catenin and the organizer-specific genes siamois and goosecoid. At low expression levels, MKK6E induces expression of lateral genes without inducing the expression of dorsal genes. Loss of CREB or p38 MAPK activity enables the expansion of the ventral homeobox gene vent1 into the dorsal marginal region, preventing the lateral expression of Xmyf5. Overall, these data indicate that dorsal-ventral mesoderm patterning is regulated by differential p38/CREB activities along the axis.
Collapse
Affiliation(s)
- Aviad Keren
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| | | | | |
Collapse
|
39
|
Misra K, Mishra K, Gui H, Matise MP. Prox1 regulates a transitory state for interneuron neurogenesis in the spinal cord. Dev Dyn 2008; 237:393-402. [PMID: 18213584 DOI: 10.1002/dvdy.21422] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Proper central nervous system (CNS) function depends critically on the generation of functionally distinct neuronal types in specific and reproducible positions. The generation of neuronal diversity during CNS development involves a fine balance between dividing neural progenitors and the differentiated neuronal progeny that they produce. However, the molecular mechanisms that regulate these processes are still poorly understood. Here, we show that the Prox1 transcription factor, which is expressed transiently and specifically in spinal interneurons, plays an important role in neurogenesis. Using both gain- and loss-of-function approaches, we find that Prox1 is capable of driving neuronal precursors out of the cell cycle and can initiate limited expression of neuronal proteins. Using RNAi approaches, we show that Prox1 function is required to execute a neurogenic differentiation program downstream of Mash1 and Ngn2. Our studies demonstrate an important, spinal interneuron-specific role for Prox1 in controlling steps required for both cell-cycle withdrawal and differentiation.
Collapse
Affiliation(s)
- Kamana Misra
- Robert Wood Johnson Medical School, UMDNJ, Piscataway, New Jersey 08854, USA
| | | | | | | |
Collapse
|
40
|
Prdm1- and Sox6-mediated transcriptional repression specifies muscle fibre type in the zebrafish embryo. EMBO Rep 2008; 9:683-9. [PMID: 18535625 PMCID: PMC2424280 DOI: 10.1038/embor.2008.73] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 03/20/2008] [Accepted: 04/02/2008] [Indexed: 11/20/2022] Open
Abstract
The zebrafish u-boot (ubo) gene encodes the transcription factor Prdm1, which is essential for the specification of the primary slow-twitch muscle fibres that derive from adaxial cells. Here, we show that Prdm1 functions by acting as a transcriptional repressor and that slow-twitch-specific muscle gene expression is activated by Prdm1-mediated repression of the transcriptional repressor Sox6. Genes encoding fast-specific isoforms of sarcomeric proteins are ectopically expressed in the adaxial cells of ubotp39 mutant embryos. By using chromatin immunoprecipitation, we show that these are direct targets of Prdm1. Thus, Prdm1 promotes slow-twitch fibre differentiation by acting as a global repressor of fast-fibre-specific genes, as well as by abrogating the repression of slow-fibre-specific genes.
Collapse
|
41
|
Ishibashi H, Matsumura N, Hanafusa H, Matsumoto K, De Robertis E, Kuroda H. Expression of Siamois and Twin in the blastula Chordin/Noggin signaling center is required for brain formation in Xenopus laevis embryos. Mech Dev 2008; 125:58-66. [PMID: 18036787 PMCID: PMC2292103 DOI: 10.1016/j.mod.2007.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 10/04/2007] [Accepted: 10/09/2007] [Indexed: 11/17/2022]
Abstract
The blastula Chordin- and Noggin-expressing (BCNE) center located in the dorsal animal region of the Xenopus blastula embryo contains both prospective anterior neuroectoderm and Spemann organizer precursor cells. Here we show that, contrary to previous reports, the canonical Wnt target homeobox genes, Double knockdown of these genes using antisense morpholinos in Xenopus laevis blocked head formation, reduced the expression of the other BCNE center genes, upregulated Bmp4 expression, and nullified hyperdorsalization by lithium chloride. Moreover, gain- and loss-of-function experiments showed that Siamois and Twin expression is repressed by the vegetal transcription factor VegT. We propose that VegT expression causes maternal beta-Catenin signals to restrict Siamois and Twin expression to the BCNE region. A two-step inhibition of BMP signals by Siamois and Twin-- first by transcriptional repression of Bmp4 and then by activation of the expression of the BMP inhibitors Chordin and Noggin--in the BCNE center is required for head formation.
Collapse
Affiliation(s)
- Hideyuki Ishibashi
- Faculty of Education (Biology), Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | - Noriko Matsumura
- Faculty of Education (Biology), Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | - Hiroshi Hanafusa
- Department of Molecular Biology, Graduate School of Science, Institute for Advanced Research, Nagoya University, Chikusa-ku, Nagoya 464-8602, Japan
- CREST, Japan Science and Technology Corporation, Chikusa-ku, Nagoya 464-8602, Japan
| | - Kunihiro Matsumoto
- Department of Molecular Biology, Graduate School of Science, Institute for Advanced Research, Nagoya University, Chikusa-ku, Nagoya 464-8602, Japan
- CREST, Japan Science and Technology Corporation, Chikusa-ku, Nagoya 464-8602, Japan
| | - E.M. De Robertis
- Howard Hughes Medical Institute and Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662, USA
| | - Hiroki Kuroda
- Faculty of Education (Biology), Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| |
Collapse
|
42
|
Vonica A, Gumbiner BM. The Xenopus Nieuwkoop center and Spemann-Mangold organizer share molecular components and a requirement for maternal Wnt activity. Dev Biol 2007; 312:90-102. [PMID: 17964564 PMCID: PMC2170525 DOI: 10.1016/j.ydbio.2007.09.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 09/06/2007] [Accepted: 09/06/2007] [Indexed: 10/22/2022]
Abstract
In Xenopus embryos, the dorso-ventral and antero-posterior axes are established by the Spemann-Mangold organizer. According to the prevalent model of early development, the organizer is induced by the dorsalizing Nieuwkoop signal, which is secreted by the Nieuwkoop center. Formation of the center requires the maternal Wnt pathway, which is active on the dorsal side of embryos. Nevertheless, the molecular nature of the Nieuwkoop signal remains unclear. Since the Nieuwkoop center and the organizer both produce dorsalizing signals in vitro, we asked if they might share molecular components. We find that vegetal explants, the source of Nieuwkoop signal in recombination assays, express a number of organizer genes. The product of one of these genes, chordin, is required for signaling, suggesting that the organizer and the center share at least some molecular components. Furthermore, experiments with whole embryos show that maternal Wnt activity is required in the organizer just as it is needed in the Nieuwkoop center in vitro. We conclude that the maternal Wnt pathway generates the Nieuwkoop center in vitro and the organizer in vivo by activating a common set of genes, without the need of an intermediary signaling step.
Collapse
Affiliation(s)
- Alin Vonica
- The Laboratory of Vertebrate Embryology, The Rockefeller University, P.O. Box 32, 1230 York Avenue, New York, NY 10021, USA
| | | |
Collapse
|
43
|
Bowes JB, Snyder KA, Segerdell E, Gibb R, Jarabek C, Noumen E, Pollet N, Vize PD. Xenbase: a Xenopus biology and genomics resource. Nucleic Acids Res 2007; 36:D761-7. [PMID: 17984085 PMCID: PMC2238855 DOI: 10.1093/nar/gkm826] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Xenbase (www.xenbase.org) is a model organism database integrating a diverse array of biological and genomic data on the frogs, Xenopus laevis and Xenopus (Silurana) tropicalis. Data is collected from other databases, high-throughput screens and the scientific literature and integrated into a number of database modules covering subjects such as community, literature, gene and genomic analysis. Gene pages are automatically assembled from data piped from the Entrez Gene, Gurdon Institute, JGI, Metazome, MGI, OMIM, PubMed, Unigene, Zfin, commercial suppliers and others. These data are then supplemented with in-house annotation. Xenbase has implemented the Gbrowse genome browser and also provides a BLAST service that allows users to specifically search either laevis or tropicalis DNA or protein targets. A table of Xenopus gene synonyms has been implemented and allows the genome, genes, publications and high-throughput gene expression data to be seamlessly integrated with other Xenopus data and to external database resources, making the wealth of developmental and functional data from the frog available to the broader research community.
Collapse
Affiliation(s)
- Jeff B Bowes
- Department of Computer Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Le TN, Du G, Fonseca M, Zhou QP, Wigle JT, Eisenstat DD. Dlx homeobox genes promote cortical interneuron migration from the basal forebrain by direct repression of the semaphorin receptor neuropilin-2. J Biol Chem 2007; 282:19071-81. [PMID: 17259176 DOI: 10.1074/jbc.m607486200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Dlx homeobox genes play an important role in vertebrate forebrain development. Dlx1/Dlx2 null mice die at birth with an abnormal cortical phenotype, including impaired differentiation and migration of GABAergic interneurons to the neocortex. However, the molecular basis for these defects downstream of loss of Dlx1/Dlx2 function is unknown. Neuropilin-2 (NRP-2) is a receptor for Class III semaphorins, which inhibit neuronal migration. Herein, we show that Neuropilin-2 is a specific DLX1 and DLX2 transcriptional target by applying chromatin immunoprecipitation to embryonic forebrain tissues. Both homeobox proteins repress Nrp-2 expression in vitro, confirming the functional significance of DLX binding. Furthermore, the homeodomain of DLX1 and DLX2 is necessary for DNA binding and this binding is essential for Dlx repression of Nrp-2 expression. Of importance, there is up-regulated and aberrant expression of NRP-2 in the forebrains of Dlx1/Dlx2 null mice. This is the first report that DLX1 or DLX2 can function as transcriptional repressors. Our data show that DLX proteins specifically mediate the repression of Neuropilin-2 in the developing forebrain. As well, our results support the hypothesis that down-regulation of Neuropilin-2 expression may facilitate tangential interneuron migration from the basal forebrain.
Collapse
Affiliation(s)
- Trung N Le
- Department of Biochemistry and Medical Genetics, Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Manitoba R3E 0V9, Canada
| | | | | | | | | | | |
Collapse
|
45
|
White JA, Heasman J. Maternal control of pattern formation inXenopus laevis. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2007; 310:73-84. [PMID: 17219372 DOI: 10.1002/jez.b.21153] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We review the essential role of maternal factors in pattern formation for Xenopus laevis, focusing on VegT, Vg1, and Wnt11. Results from loss of function experiments demonstrate a clear requirement for these genes in germ layer specification, dorsal-ventral axis formation, and convergence extension. We also discuss these genes in the broader context of metazoan development, exploring whether and how their functions in the X. laevis model organism may or may not be conserved in other species. Wnt11 signaling in particular provides a classic example where understanding context in development is crucial to understanding function. Genomic sequencing, gene expression, and functional screening data that are becoming available in more species are providing invaluable aid to decoding and modeling signaling pathways. More work is needed to develop a comprehensive catalog of the Wnt signaling, T-box, and TGF-beta genes in metazoans both near and far in evolutionary distance. We finally discuss some specific experimental and modeling efforts that will be needed to understand the behavior of these signaling networks in vivo so that we can interpret these critical pathways in an evolutionary framework.
Collapse
Affiliation(s)
- Jody A White
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio 45229-3039, USA.
| | | |
Collapse
|
46
|
Steiner AB, Engleka MJ, Lu Q, Piwarzyk EC, Yaklichkin S, Lefebvre JL, Walters JW, Pineda-Salgado L, Labosky PA, Kessler DS. FoxD3 regulation of Nodal in the Spemann organizer is essential for Xenopus dorsal mesoderm development. Development 2006; 133:4827-38. [PMID: 17092955 PMCID: PMC1676154 DOI: 10.1242/dev.02663] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Induction and patterning of the mesodermal germ layer is a key early step of vertebrate embryogenesis. We report that FoxD3 function in the Xenopus gastrula is essential for dorsal mesodermal development and for Nodal expression in the Spemann organizer. In embryos and explants, FoxD3 induced mesodermal genes, convergent extension movements and differentiation of axial tissues. Engrailed-FoxD3, but not VP16-FoxD3, was identical to native FoxD3 in mesoderm-inducing activity, indicating that FoxD3 functions as a transcriptional repressor to induce mesoderm. Antagonism of FoxD3 with VP16-FoxD3 or morpholino-knockdown of FoxD3 protein resulted in a complete block to axis formation, a loss of mesodermal gene expression, and an absence of axial mesoderm, indicating that transcriptional repression by FoxD3 is required for mesodermal development. FoxD3 induced mesoderm in a non-cell-autonomous manner, indicating a role for secreted inducing factors in the response to FoxD3. Consistent with this mechanism, FoxD3 was necessary and sufficient for the expression of multiple Nodal-related genes, and inhibitors of Nodal signaling blocked mesoderm induction by FoxD3. Therefore, FoxD3 is required for Nodal expression in the Spemann organizer and this function is essential for dorsal mesoderm formation.
Collapse
Affiliation(s)
- Aaron B. Steiner
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Mark J. Engleka
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Qun Lu
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Eileen C. Piwarzyk
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Sergey Yaklichkin
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Julie L. Lefebvre
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - James W. Walters
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Liliam Pineda-Salgado
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | | | | |
Collapse
|
47
|
Martins GA, Hutchins AS, Reiner SL. Transcriptional activators of helper T cell fate are required for establishment but not maintenance of signature cytokine expression. THE JOURNAL OF IMMUNOLOGY 2005; 175:5981-5. [PMID: 16237092 DOI: 10.4049/jimmunol.175.9.5981] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The stability of helper T cell fates is not well understood. Using conditional introduction of dominant-negative factors, we now show that T-bet and GATA-3 are far more critical in establishment than maintenance of IFN-gamma and IL-4 activity during Th1 and Th2 maturation, respectively. We also show that a genetic interaction between T-bet and its target Hlx seems to be required for Th1 maturation, but that Hlx may also be dispensable for maintenance of a transcriptionally permissive ifng gene. In parallel to progressive activator independence in the permissive lineage, the ifng gene becomes more recalcitrant to switching as the forbidden lineage matures. T-bet plus Hlx can disrupt ifng silencing when introduced into developing Th2 cells, but they fail to perturb ifng silencing in mature Th2 cells. In contrast, a hypermorphic allele of T-bet can reverse silencing of the ifng gene in mature Th2 cells. These results suggest that signature gene activity of helper T cells is initially plastic but later becomes epigenetically fixed and offer an initial strategy for inducing mature cells to switch their fate.
Collapse
Affiliation(s)
- Gislâine A Martins
- Abramson Family Cancer Research Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
48
|
Wilson KA, Andrews ME, Rudolf Turner F, Raff RA. Major regulatory factors in the evolution of development: the roles of goosecoid and Msx in the evolution of the direct-developing sea urchin Heliocidaris erythrogramma. Evol Dev 2005; 7:416-28. [PMID: 16174035 DOI: 10.1111/j.1525-142x.2005.05046.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The transcription factors Gsc and Msx are expressed in the oral ectoderm of the indirect-developing sea urchin Heliocidaris tuberculata. Their patterns of expression are highly modified in the direct developer Heliocidaris erythrogramma, which lacks an oral ectoderm. We here test the hypothesis that they are large effect genes responsible for the loss of the oral ectoderm module in the direct-developing larva of H. erythrogramma as well as for the restoration of an overt oral ectoderm in H.e. xH.t. hybrids. We undertook misexpression/overexpression and knockdown assays in the two species and in hybrids by mRNA injection. The results indicate that dramatic changes of function of these transcription factors has occurred. One of these genes, Gsc, has the ability when misexpressed to partially restore oral ectoderm in H. erythrogramma. On the other hand, Msx has lost any oral function and instead has a role in mesoderm proliferation and patterning. In addition, we found that the H. tuberculataGsc is up regulated in H.e. xH.t. hybrids, showing a preferential use of the indirect developing parental gene in the development of the hybrid. We suggest that Gsc qualifies as a gene of large evolutionary effect and is partially responsible for the evolution of direct development of H. erythrogramma. We discuss these results in light of modularity and genetic networks in development, as well as in their implications for the rapid evolution of large morphological changes in development.
Collapse
Affiliation(s)
- Keen A Wilson
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | | |
Collapse
|
49
|
Park JI, Kim SW, Lyons JP, Ji H, Nguyen TT, Cho K, Barton MC, Deroo T, Vleminckx K, Moon RT, McCrea PD. Kaiso/p120-catenin and TCF/beta-catenin complexes coordinately regulate canonical Wnt gene targets. Dev Cell 2005; 8:843-54. [PMID: 15935774 DOI: 10.1016/j.devcel.2005.04.010] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Revised: 03/01/2005] [Accepted: 04/16/2005] [Indexed: 11/18/2022]
Abstract
Beta-catenin-dependent or canonical Wnt signals are fundamental in animal development and tumor progression. Using Xenopus laevis, we report that the BTB/POZ zinc finger family member Kaiso directly represses canonical Wnt gene targets (Siamois, c-Fos, Cyclin-D1, and c-Myc) in conjunction with TCF/LEF (TCF). Analogous to beta-catenin relief of TCF repressive activity, we show that p120-catenin relieves Kaiso-mediated repression of Siamois. Furthermore, Kaiso and TCF coassociate, and combined Kaiso and TCF derepression results in pronounced Siamois expression and increased beta-catenin coprecipitation with the Siamois promoter. The functional interdependency is underlined by Kaiso suppression of beta-catenin-induced axis duplication and by TCF-3 rescue of Kaiso depletion phenotypes. These studies point to convergence of parallel p120-catenin/Kaiso and beta-catenin/TCF signaling pathways to regulate gene expression in vertebrate development and possibly carcinogenesis.
Collapse
Affiliation(s)
- Jae-Il Park
- Department of Biochemistry and Molecular Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Siamois is the transcriptional mediator of the dorsal Wnt signaling pathway and is necessary for formation of the Spemann organizer and dorsoanterior development in Xenopus. We have determined that XIC, a Xenopus I-mfa domain protein that regulates Tcf3 binding, is required for dorsoaxial development and specifically for Siamois activity in establishing the dorsal organizer. In loss-of-function studies, we found that embryos injected with a morpholino to XIC mRNA (XIC morphpolino) are missing head structures, neural tube, notochord, and paraxial mesoderm as well as NCAM and XMyoD expression. Although Siamois, Twin, and Xnr3 expression is normal in morpholino-injected embryos, levels of downstream organizer factors, including goosecoid, Xnot, Cerberus, and chordin, are severely reduced. Ectopic axis formation induced by Siamois is repressed by injection of the XIC morpholino and further repressed by coinjection of beta-catenin or a constitutively active Tcf3/HMG/G4A fusion. Activation of reporters driven by the Siamois-responsive proximal element of the goosecoid promoter is inhibited in the presence of the morpholino and can be rescued by murine I-mfa and by a dominant-negative Tcf3. The data indicate a role for XIC in limiting Tcf3-dependent repression of Siamois activities that are required for goosecoid transcription and for dorsal organizer formation.
Collapse
Affiliation(s)
- Lauren Snider
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | | |
Collapse
|